Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Neural Transm (Vienna) ; 127(12): 1631-1640, 2020 12.
Article in English | MEDLINE | ID: mdl-32778969

ABSTRACT

5'-Nucleotidase domain-containing protein 2 (NT5DC2) has been revealed by genome-wide association studies (GWAS) as a gene implicated in neuropsychiatric disorders related to the abnormality of dopamine (DA) activity in the brain. Based on its amino acid sequence, NT5DC2 is assumed to be a member of the family of haloacid dehalogenase-type phosphatases; although there is no information about its function and structural conformation. We recently reported that NT5DC2 binds to tyrosine hydroxylase (TH) and that the down-regulation of NT5DC2 tended to increase DA synthesis. In this study, we investigated whether NT5DC2 could regulate the catalytic activity of TH, which converts tyrosine to DOPA, because the phosphorylation level of TH, controlled by protein kinases and phosphatases, is well known to regulate its catalytic activity. The down-regulation of NT5DC2 by siRNA increased mainly DOPA synthesis by TH in PC12D cells, although this down-regulation tended to increase the conversion of DOPA to DA by aromatic L-amino acid decarboxylase. The increased DOPA synthesis should be attributed to the catalytic activity of TH controlled by its phosphorylation, because Western blot analysis revealed that the down-regulation of NT5DC2 tended to increase the level of TH phosphorylated at its Ser residues, but not that of the TH protein. Moreover, the induction of kinase activity by forskolin markedly potentiated the phosphorylation of TH at its Ser40 in PC12D cells having down-regulated NT5DC2. Immunocytochemical analysis of PC12D cells demonstrated that NT5DC2, TH protein, and TH phosphorylated at its Ser40 were predominantly localized in the cytoplasm and that the localization of NT5DC2 and TH proteins partially overlapped. Collectively, our results indicate that NT5DC2 could work to inhibit the DOPA synthesis by decreasing the phosphorylation of TH at its Ser40. We propose that NT5DC2 might decrease this phosphorylation of TH by promoting dephosphorylation or by inhibiting kinase activity.


Subject(s)
Genome-Wide Association Study , Tyrosine 3-Monooxygenase , Dopamine , Phosphorylation , Tyrosine , Tyrosine 3-Monooxygenase/metabolism
2.
Biochem Biophys Res Commun ; 516(4): 1060-1065, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31279527

ABSTRACT

Tyrosine hydroxylase (TH), which catalyzes the conversion of l-tyrosine to l-DOPA, is the rate-limiting enzyme in the biosynthesis of catecholamines. It is well known that both α-synuclein and 14-3-3 protein family members bind to the TH molecule and regulate phosphorylation of its N-terminus by kinases to control the catalytic activity. In this present study we investigated whether other proteins aside from these 2 proteins might also bind to TH molecules. Nano-LC-MS/MS analysis revealed that 5'-nucleotidase domain-containing protein 2 (NT5DC2), belonging to a family of haloacid dehalogenase-type (HAD) phosphatases, was detected in the immunoprecipitate of PC12D cell lysates that had been reacted with Dynabeads protein G-anti-TH antibody conjugate. Surprisingly, NT5DC2 had already been revealed by Genome-Wide Association Studies (GWAS) as a gene implicated in neuropsychiatric disorders such as schizophrenia, bipolar disorder, which are diseases related to the abnormality of dopamine activity in the brain, although the role that NT5DC2 plays in these diseases remains unknown. Therefore, we investigated the effect of NT5DC2 on the TH molecule. The down-regulation of NT5DC2 by siRNA increased the synthesis of catecholamines (dopamine, noradrenaline, and adrenaline) in PC12D cells. These increases might be attributed to the catalytic activity of TH and not to the intracellular stability of TH, because the intracellular content of TH assessed by Western blotting was not changed by the down-regulation of NT5DC2. Collectively, our results indicate that NT5DC2 inhibited the synthesis of dopamine by decreasing the enzymatic activity of TH.


Subject(s)
5'-Nucleotidase/metabolism , Catecholamines/metabolism , Tyrosine 3-Monooxygenase/metabolism , 5'-Nucleotidase/genetics , Animals , Cell Line , Chromatography, Liquid , Down-Regulation , PC12 Cells , Protein Binding , RNA Interference , RNA, Small Interfering/genetics , Rats , Tandem Mass Spectrometry
3.
J Neural Transm (Vienna) ; 125(1): 9-15, 2018 01.
Article in English | MEDLINE | ID: mdl-27866280

ABSTRACT

Tyrosine hydroxylase (TH) is the rate-limiting enzyme in catecholamine biosynthesis, and its stability is a fundamental factor to maintain the level of the catecholamines in cells. However, the intracellular stability of TH determined by the degradation remains unknown; although the TH molecule phosphorylated at its Ser19 was observed in the nucleus, and the phosphorylation suspected to trigger its proteasome-mediated degradation. Computer-assisted analysis using the cNLS Mapper program predicted that two sequences of nuclear localization signals (NLS) exist in the N-terminus of TH molecule containing the phosphorylation sites Ser19, Ser31, and Ser40 (Pro9-Arg38 and Lys12-Ile42): the NLS scores indicated that TH could become localized in both nucleus and cytoplasm. Moreover, inhibition of the importin α/ß-mediated nuclear import pathway increased the level of TH phosphorylated at its Ser19 in PC12D cells. The results suggest that TH might be imported to nucleus from cytoplasm to be degraded. Recent studies revealed that proteasomes predominantly exist in the nucleus rather than in the cytoplasm to degrade the nuclear proteins related to cell-cycle progression, gene expression, DNA damage, and DNA repair. Therefore, these studies suggest that the relationship between the phosphorylation and the nuclear localization of the TH molecule should be a matter of focus to understand the mechanism of proteasome-mediated degradation of the enzyme as a first priority.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Proteasome Endopeptidase Complex/metabolism , Tyrosine 3-Monooxygenase/metabolism , Amino Acid Sequence , Animals , Cell Nucleus/chemistry , Cytoplasm/chemistry , Humans , Intracellular Fluid/chemistry , Intracellular Fluid/metabolism , Phosphorylation/physiology , Proteasome Endopeptidase Complex/analysis , Tyrosine 3-Monooxygenase/analysis
4.
Biochem Biophys Res Commun ; 472(4): 598-602, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26969276

ABSTRACT

Tyrosine hydroxylase (TH) is the rate-limiting enzyme in catecholamine biosynthesis, and its stability is a fundamental factor to maintain the level of the catecholamines in cells. However, the intracellular stability determined by the degradation pathway remains unknown. In this study, we investigated the mechanism by which phosphorylation of TH affected the proteasome pathway. The inhibition of proteasomes by MG-132 increased the percentage of TH molecules phosphorylated at their Ser19, Ser31 and/or Ser40 among the total TH proteins to about 70% in PC12D cells over a 24-hr period; although the percentage of phosphorylated TH molecules was about 20% under basal conditions. Moreover, the inhibition of proteasomes by epoxomicin with high specificity increased primarily the quantity of TH molecules phosphorylated at their Ser19. The phosphorylation of Ser19 potentiated Ser40 phosphorylation in cells by a process known as hierarchical phosphorylation. Therefore, the proteasome inhibition might result in an increase in the levels of all 3 phosphorylated TH forms, thus complicating interpretation of data. Conversely, activation of proteasome degradation by IU-1, which is an inhibitor for the deubiquitinating activity of USP14, decreased only the quantity of TH molecules phosphorylated at their Ser19, although it did not decrease that of TH phosphorylated at its Ser31 and Ser40 or that of TH molecules. These results suggest that the phosphorylation of Ser19 in the N-terminal portion of TH is critical as a trigger for the degradation of this enzyme by the ubiquitin-proteasome pathway.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , PC12 Cells , Phosphorylation , Proteolysis , Rats , Signal Transduction , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitination
5.
J Neural Transm (Vienna) ; 122(2): 187-99, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24919883

ABSTRACT

We previously reported that an optimal dose of lipopolysaccharide (LPS) markedly extends the lifespan of murine primary-cultured microglia by suppressing cell death pathways. In this study, we investigated the effects of LPS pretreatment on UV light-induced apoptosis of cells from the microglial cell line BV-2. More than half of BV-2 cells were apoptotic, and procaspase-3 was cleaved into its active form at 3 h of UV irradiation. In contrast, in BV-2 cells treated with LPS for 24 h, UV irradiation caused neither apoptosis nor procaspase-3 cleavage. LPS treatment arrested the cell cycle in G1 phase and upregulated cyclin-dependent kinase inhibitor p21(Waf1/Cip1) and growth arrest and DNA damage-inducible (GADD) 45α in BV-2 cells. When p21(Waf1/Cip1) and GADD45α were knocked down by small interfering RNA, procaspase-3 was cleaved into its active form to induce apoptosis. Our findings suggest that LPS inhibits UV-induced apoptosis in BV-2 cells through arrest of the cell cycle in G1 phase by upregulation of p21(Waf1/Cip1) and GADD45α. Excessive activation of microglia may play a critical role in the exacerbation of neurodegeneration, therefore, normalizing the precise regulation of apoptosis may be a new strategy to prevent the deterioration caused by neurodegenerative disorders.


Subject(s)
Apoptosis/drug effects , G1 Phase/drug effects , Lipopolysaccharides/pharmacology , Microglia/drug effects , Ultraviolet Rays , Animals , Apoptosis/radiation effects , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/genetics , Cyclins/metabolism , Flow Cytometry , G1 Phase/radiation effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Mice , Microglia/radiation effects , Oligonucleotide Array Sequence Analysis , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger , RNA, Small Interfering/pharmacology , Time Factors
6.
J Neural Transm (Vienna) ; 122(6): 757-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25504008

ABSTRACT

We previously showed that aripiprazole increases intracellular NADPH and glucose-6-phosphate dehydrogenase mRNA in PC12 cells. Aripiprazole presumably activates a system that concurrently detoxifies reactive oxygen species and replenishes NADPH. Nrf2, a master transcriptional regulator of redox homeostasis genes, also activates the pentose phosphate pathway, including NADPH production. Therefore, our aim was to determine whether aripiprazole activates Nrf2 in PC12 cells. Aripiprazole increased mRNA expression of Nrf2-dependent genes (NAD(P)H-quinone oxidoreductase-1, Nqo1; heme oxygenase-1, HO1; and glutamate-cysteine ligase catalytic subunit) and protein expression of Nqo1 and HO1 in these cells (p < 0.05). To maintain increased Nrf2 activity, it is necessary to inhibit Nrf2 degradation; this is done by causing Nrf2 to dissociate from Keap1 or ß-TrCP. However, in aripiprazole-treated cells, the relative amount of Nrf2 anchored to Keap1 or ß-TrCP was unaffected and Nrf2 in the nuclear fraction decreased (p < 0.05). Aripiprazole did not affect phosphorylation of Nrf2 at Ser40 and decreased the relative amount of acetylated Nrf2 (p < 0.05). The increase in Nqo1 and HO1 in aripiprazole-treated cells cannot be explained by the canonical Nrf2-degrading pathways. Further experiments are needed to determine the biochemical mechanisms underlying the aripiprazole-induced increase in these enzymes.


Subject(s)
Antipsychotic Agents/pharmacology , Aripiprazole/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Acetylation/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cell Survival/drug effects , Cytosol/drug effects , Cytosol/enzymology , Glutamate-Cysteine Ligase/metabolism , Hydrogen Peroxide/toxicity , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , PC12 Cells , Phosphorylation/drug effects , Rats , beta-Transducin Repeat-Containing Proteins/metabolism
7.
J Neural Transm (Vienna) ; 121(1): 91-103, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23934573

ABSTRACT

In aripiprazole-treated PC12 cells, we previously showed that the mitochondrial membrane potential (Δψm) was rather increased in spite of lowered cytochrome c oxidase activity. To address these inconsistent results, we focused the NADPH generation by glucose-6-phosphate dehydrogenase (G6PD), a rate-limiting enzyme of the pentose phosphate pathway (PPP), to titrate reactive oxygen species (ROS) that results in the Δψm maintenance. G6PD may be also involved in another inconsistent result of lowered intracellular lactate level in aripiprazole-treated PC12 cells, because PPP competes glucose-6-phosphate with the glycolytic pathway, resulting in the downregulation of glycolysis. Therefore, we assayed intracellular amounts of NADPH, ROS, and the activities of the enzymes generating or consuming NADPH (G6PD, NADP(+)-dependent isocitrate dehydrogenase, NADP(+)-dependent malic enzyme, glutathione reductase, and NADPH oxidase [NOX]) and estimated glycolysis in 50 µM aripiprazole-, clozapine-, and haloperidol-treated PC12 cells. NADPH levels were enhanced only in aripiprazole-treated ones. Only haloperidol increased ROS. However, the enzyme activities did not show significant changes toward enhancing NADPH level except for the aripiprazole-induced decrease in NOX activity. Thus, the lowered NOX activity could have contributed to the aripiprazole-induced increase in the NADPH level by lowering ROS generation, resulting in maintained Δψm. Although the aforementioned assumption was invalid, the ratio of fructose-1,6-bisphosphate to fructose-6-phosphate was decreased by all antipsychotics examined. Pyruvate kinase activity was enhanced only by aripiprazole. In summary, these observations indicate that aripiprazole possibly possesses the pharmacological superiority to clozapine and haloperidol in the ROS generation and the adjustment of glycolytic pathway.


Subject(s)
Antipsychotic Agents/pharmacology , NADPH Oxidases/metabolism , NADP/metabolism , Neurons/drug effects , Piperazines/pharmacology , Quinolones/pharmacology , Animals , Aripiprazole , Neurons/metabolism , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
8.
J Neural Transm (Vienna) ; 120(1): 49-54, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22644539

ABSTRACT

Postmortem brain biochemistry has revealed that the main symptom of movement disorder in Parkinson's disease (PD) is caused by a deficiency in dopamine (DA) at the nerve terminals of degenerating nigro-striatal DA neurons in the striatum. Since tyrosine hydroxylase (TH) is the rate-limiting enzyme for the biosynthesis of DA, TH may play an important role in the disease process of PD. DA regulated by TH activity is thought to interact with α-synuclein protein, which results in intracellular aggregates called Lewy bodies and causes apoptotic cell death during the aging process. Human TH has several isoforms produced by alternative mRNA splicing, which may affect activation by phosphorylation of serine residues in the N-terminus of TH. The activity and protein level of TH are decreased to cause DA deficiency in the striatum in PD. However, the homo-specific activity (activity/enzyme protein) of TH is increased. This increase in TH homo-specific activity suggests activation by increased phosphorylation at the N-terminus of the TH protein for a compensatory increase in DA synthesis. We recently found that phosphorylation of the N-terminal portion of TH triggers proteasomal degradation of the enzyme to increase TH turnover. We propose a hypothesis that this compensatory activation of TH by phosphorylation in the remaining DA neurons may contribute to a further decrease in TH protein and activity in DA neurons in PD, causing a vicious circle of decreasing TH activity, protein level and DA contents. Furthermore, increased TH homo-specific activity leading to an increase in DA may cause toxic reactive oxygen species in the neurons to promote neurodegeneration.


Subject(s)
Brain/enzymology , Parkinson Disease/pathology , Tyrosine 3-Monooxygenase/metabolism , Brain/pathology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Postmortem Changes
9.
Cell Mol Neurobiol ; 32(5): 777-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22189676

ABSTRACT

This review summarizes the effects of neuroinflammatory stress on the subventricular zone (SVZ), where new neurons are constitutively produced in the adult brain, especially focusing on the relation with Parkinson's disease (PD), because the SVZ is under the control of dopaminergic afferents from the substantia nigra (SN). In Lewy bodies-positive-PD, microglia is known to phagocytoze aggregated α-synuclein, resulting in the release of inflammatory cytokines. The neurogenesis in the SVZ should be affected in PD brain by the neuroinflammatory process. The administration of lipopolysaccaharide is available as an alternative model for microglia-induced loss of dopaminergic neurons and also the impairment of stem cell maintenance. Therefore, the research on the neuroinflammatory process in the SVZ gives us a hint to prevent the outbreak of PD or at least slow the disease process.


Subject(s)
Cerebral Ventricles/pathology , Inflammation/pathology , Nervous System/pathology , Parkinson Disease/pathology , Stress, Physiological , Animals , Humans , Signal Transduction
10.
J Neural Transm (Vienna) ; 119(11): 1327-42, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22392058

ABSTRACT

Aripiprazole is the only atypical antipsychotic drug known to cause the phosphorylation of AMP-activated protein kinase (AMPK) in PC12 cells. However, the molecular mechanisms underlying this phosphorylation in aripiprazole-treated PC12 cells have not yet been clarified. Here, using PC12 cells, we show that these cells incubated for 24 h with aripiprazole at 50 µM and 25 mM glucose underwent a decrease in their NAD⁺/NADH ratio. Aripiprazole suppressed cytochrome c oxidase (COX) activity but enhanced the activities of pyruvate dehydrogenase (PDH), citrate synthase and Complex I. The changes in enzyme activities coincided well with those in NADH, NAD⁺, and NAD⁺/NADH ratio. However, the bioenergetic peril judged by the lowered COX activity might not be accompanied by excessive occurrence of apoptotic cell death in aripiprazole-treated cells, because the mitochondrial membrane potential was not decreased, but rather increased. On the other hand, when PC12 cells were incubated for 24 h with clozapine at 50 µM and 25 mM glucose, the NAD⁺/NADH ratio did not change. Also, the COX activity was decreased; and the PDH activity was enhanced. These results suggest that aripiprazole-treated PC12 cells responded to the bioenergetic peril more effectively than the clozapine-treated ones to return the ATP biosynthesis back toward its ordinary level. This finding might be related to the fact that aripiprazole alone causes phosphorylation of AMPK in PC12 cells.


Subject(s)
Antipsychotic Agents/pharmacology , Carbon/metabolism , Clozapine/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glycolysis/drug effects , Piperazines/pharmacology , Quinolones/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Aripiprazole , Cell Survival/drug effects , Dihydrolipoamide Dehydrogenase/genetics , Dihydrolipoamide Dehydrogenase/metabolism , Dose-Response Relationship, Drug , Electron Transport Complex IV/metabolism , Extracellular Fluid/drug effects , Glucose/pharmacology , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Ketone Oxidoreductases/genetics , Ketone Oxidoreductases/metabolism , Lactic Acid/metabolism , Membrane Potential, Mitochondrial/drug effects , NAD/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , PC12 Cells/drug effects , PC12 Cells/enzymology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvic Acid/metabolism , RNA, Messenger/metabolism , Rats , Time Factors
11.
Clin Exp Pharmacol Physiol ; 39(7): 599-607, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22519637

ABSTRACT

1. Previously, we reported that an optimal dose of lipopolysaccharide (LPS) markedly extends the life span of mouse primary-cultured microglia by suppressing apoptotic and autophagic cell death pathways. The aim of the present study was to assess how these cells protect themselves against reactive oxygen species (ROS) generated by LPS treatment. 2. The study was conducted in microglia obtained from murine neonate brain, which are destined to die within a few days under ordinary culture conditions. 3. The generation of ROS was maximal after 15 h LPS treatment (1 ng/mL LPS and 100 ng/mL LPS). The expression of inducible nitric oxide (NO) synthase protein was significantly increased by Day 1 of LPS treatment and was followed by the production of NO. The expression of either Cu/Zn- or Mn-superoxide dismutase protein (SOD) was also increased by 16 h and Day 1 of LPS treatment. LPS did not affect the expression of Cu/Zn- and Mn-SOD proteins, nor did it extend the life span of microglia that had mutated Toll-like receptor (TLR) 4. 4. The findings of the present study suggest that SODs function as a potent barrier to overcome ROS generated in primary-cultured microglia following LPS treatment and that TLR4 may be significantly involved in inducing these proteins. The microglia may be able to protect themselves against oxidative stress, allowing them to live for more than 1 month. Because long-lived microglia may play a critical role in the exacerbation of neurodegeneration, bringing activated microglia back to their resting stage could be a new and promising strategy to inhibit the deterioration underlying neurodegenerative disorders.


Subject(s)
Free Radicals/metabolism , Microglia/metabolism , Oxidative Stress , Superoxide Dismutase/metabolism , Animals , Catalase/metabolism , Cell Separation , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C3H , Microglia/cytology , Microglia/drug effects , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase-1 , Toll-Like Receptor 4/metabolism
12.
Neurodegener Dis ; 10(1-4): 100-3, 2012.
Article in English | MEDLINE | ID: mdl-22301667

ABSTRACT

BACKGROUND: Activated microglia secrete inflammatory cytokines and may play roles in the progression of neurodegenerative diseases. However, the mechanism underlying microglial activation remains unclear. OBJECTIVE: Our aim was to examine the regulation of activated microglia through their cell death and survival pathways. METHODS: We used mouse primary-cultured microglia, which are destined to die within a few days under ordinary culture conditions. The microglia live for longer than 1 month, without any measurable increase in apoptotic or necrotic cell death, when kept activated by sublethal concentrations of lipopolysaccharide (LPS). RESULTS: LPS-treated microglia showed changes in shape. LPS treatment had no effect on the level of the proapoptotic Bcl-2-associated X protein but increased the level of the antiapoptotic protein Bcl-xL at day 1. Furthermore, the level of microtubule-associated light chain 3-II, a marker protein for autophagy, was decreased 3 h after exposure to LPS. CONCLUSION: An increase in Bcl-xL seems to inhibit both apoptosis and autophagy. Our results suggest that long-lived microglia resulting from exposure to the optimal dose of LPS may play critical roles in the progression of neurodegeneration.


Subject(s)
Apoptosis/physiology , Cytokines/metabolism , Microglia/drug effects , Animals , Apoptosis/drug effects , Brain/cytology , Caspase 3/metabolism , Cells, Cultured , Lipopolysaccharides/pharmacology , Mice , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
13.
eNeuro ; 9(2)2022.
Article in English | MEDLINE | ID: mdl-35437265

ABSTRACT

Hypothalamic melanin-concentrating hormone (MCH) neurons are important regulators of multiple physiological processes, such as sleep, feeding, and memory. Despite the increasing interest in their neuronal functions, the molecular mechanism underlying MCH neuron development remains poorly understood. We report that a three-dimensional culture of mouse embryonic stem cells (mESCs) can generate hypothalamic-like tissues containing MCH-positive neurons, which reproduce morphologic maturation, neuronal connectivity, and neuropeptide/neurotransmitter phenotype of native MCH neurons. Using this in vitro system, we demonstrate that Hedgehog (Hh) signaling serves to produce major neurochemical subtypes of MCH neurons characterized by the presence or absence of cocaine- and amphetamine-regulated transcript (CART). Without exogenous Hh signals, mESCs initially differentiated into dorsal hypothalamic/prethalamic progenitors and finally into MCH+CART+ neurons through a specific intermediate progenitor state. Conversely, activation of the Hh pathway specified ventral hypothalamic progenitors that generate both MCH+CART- and MCH+CART+ neurons. These results suggest that in vivo MCH neurons may originate from multiple cell lineages that arise through early dorsoventral patterning of the hypothalamus. Additionally, we found that Hh signaling supports the differentiation of mESCs into orexin/hypocretin neurons, a well-defined cell group intermingled with MCH neurons in the lateral hypothalamic area (LHA). The present study highlights and improves the utility of mESC culture in the analysis of the developmental programs of specific hypothalamic cell types.


Subject(s)
Hypothalamic Hormones , Mouse Embryonic Stem Cells , Animals , Hedgehog Proteins/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Melanins/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/physiology , Orexins/metabolism , Pituitary Hormones/metabolism
14.
PLoS One ; 17(11): e0276694, 2022.
Article in English | MEDLINE | ID: mdl-36356043

ABSTRACT

The hypothalamus is comprised of heterogenous cell populations and includes highly complex neural circuits that regulate the autonomic nerve system. Its dysfunction therefore results in severe endocrine disorders. Although recent experiments have been conducted for in vitro organogenesis of hypothalamic neurons from embryonic stem (ES) or induced pluripotent stem (iPS) cells, whether these stem cell-derived hypothalamic neurons can be useful for regenerative medicine remains unclear. We therefore performed orthotopic transplantation of mouse ES cell (mESC)-derived hypothalamic neurons into adult mouse brains. We generated electrophysiologically functional hypothalamic neurons from mESCs and transplanted them into the supraoptic nucleus of mice. Grafts extended their axons along hypothalamic nerve bundles in host brain, and some of them even projected into the posterior pituitary (PPit), which consists of distal axons of the magnocellular neurons located in hypothalamic supraoptic and paraventricular nuclei. The axonal projections to the PPit were not observed when the mESC-derived hypothalamic neurons were ectopically transplanted into the substantia nigra reticular part. These findings suggest that our stem cell-based orthotopic transplantation approach might contribute to the establishment of regenerative medicine for hypothalamic and pituitary disorders.


Subject(s)
Hypothalamus , Mouse Embryonic Stem Cells , Animals , Mice , Hypothalamus/physiology , Axons/physiology , Neurons/physiology , Supraoptic Nucleus , Paraventricular Hypothalamic Nucleus
15.
Biochem Biophys Res Commun ; 407(2): 343-7, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21392500

ABSTRACT

Tyrosine hydroxylase (TH) is the rate-limiting enzyme in catecholamine biosynthesis, and its N-terminus plays a critical role in the intracellular stability of the enzyme. In the present study, we investigated the mechanism by which the N-terminal region of TH affects this stability. TH molecules phosphorylated at their Ser31 and Ser40 were localized predominantly in the cytoplasm of PC12D cells. However, those molecules phosphorylated at Ser19 were found mainly in the nucleus, whereas they seemed to be negligible in the cytoplasm. The inhibition of proteasomes increased the quantity of TH molecules phosphorylated at their Ser19 and Ser40, although it did not increase that of TH molecules or that of TH phosphorylated at its Ser31. The inhibition of autophagy did not affect the amount of the TH molecule or that of its three phosphorylated forms. Deletion mutants of human TH type-1 lacking the N-terminal region containing the three phosphorylation sites possessed high stability of the enzyme in PC12D cells. These results suggest that the phosphorylation of the N-terminal portion of TH regulates the degradation of this enzyme by the ubiquitin-proteasome pathway.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin/metabolism , Animals , Autophagy , Humans , Lysosomes/enzymology , PC12 Cells , Phosphorylation , Protein Structure, Tertiary/genetics , Rats , Sequence Deletion , Tyrosine 3-Monooxygenase/genetics
16.
J Neural Transm (Vienna) ; 117(10): 1139-53, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20686905

ABSTRACT

By converting changes in intracellular energy status to changes in cell membrane polarization, ATP-sensitive K(+) (K(ATP)) channels in hypothalamic appetite-regulating neurons play a critical role in linking neuronal electrochemical function, metabolic and energy status, and feeding behavior. Most atypical antipsychotics (AAPs) increase the appetite of patients with schizophrenia and thus cause obesity. This study aimed to explain the mechanism underlying AAP-induced appetite stimulation, based on the fact that the efficiency of fatty acid uptake into mitochondria generating ATP through ß-oxidation is determined by the rate of fatty acid synthesis. Using PC12 cells exposed to clozapine, olanzapine, risperidone, quetiapine, ziprasidone, aripiprazole, and haloperidol, we measured intracellular ATP and mRNA and protein expression of enzymes and related substances involved in fatty acid synthesis and K(ATP) channel function. Forty-eight-hour treatment of cells with 50 µM aripiprazole in 5.6 mM glucose decreased intracellular ATP. Only 50 µM aripiprazole phosphorylated AMP-activated protein kinase (AMPK); none of the other antipsychotics did so to a detectable level. Expression of carnitine palmitoyltransferase 1a, uncoupling protein 2, and sulfonylurea receptor 1 was unaffected by the antipsychotics, although expression of their mRNA was affected by AAPs. Pyrilamine (H(1) receptor antagonist), ketanserin (5HT(2) receptor antagonist), and raclopride (D(2) receptor antagonist) alone or in combination had no effect on expression of the aforementioned proteins. Therefore, although this study did not differentiate orexigenic and non-orexigenic AAPs, it suggests that aripiprazole is unique in its ability to activate AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antipsychotic Agents/pharmacology , Haloperidol/pharmacology , Piperazines/pharmacology , Quinolones/pharmacology , Animals , Appetite Regulation/drug effects , Appetite Regulation/physiology , Aripiprazole , Fatty Acids/biosynthesis , Oxidative Phosphorylation/drug effects , PC12 Cells , Potassium Channels/drug effects , Potassium Channels/metabolism , RNA, Messenger/analysis , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats
17.
Neurosci Lett ; 412(3): 254-8, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17174476

ABSTRACT

Electrophysiological and immunohistochemical studies have demonstrated that glucose-sensing neurons in the hypothalamus contain both ATP-sensitive K(+) (K(ATP)) and tandem-pore K(+) (TASK1 and TASK3) channels and that glucose-induced depolarization or hyperpolarization of these neurons function as an important link between glucose-excited or glucose-inhibited neurons and feeding behavior. Medication with atypical antipsychotics increases the appetite of schizophrenic patients and thus causes increases in body weight. Therefore, the present study investigates mRNA expression levels of the genes encoding the components of these K(+) channel subsets in PC12 cells cultured with risperidone (an atypical antipsychotic) and in the hypothalami of rats subcutaneously injected for 21 consecutive days with 0.1 or 0.01 mg/kg/day of risperidone. The mRNA expression levels of various genes were not obviously altered in rat hypothalami. However, the mRNA expression levels for sulfonylurea receptor 1, a component affording nucleotide-binding folds to K(ATP) channels, and TASK1 were down-regulated in PC12 cells cultured with 50 microM risperidone for 24h, but the amount of intracellular ATP in these cells was not affected by the drug. Collectively, these results indicate that the amplitude of the current through these K(+) channels in PC12 cells might be modulated as a pharmacological effect of risperidone.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Dopamine Antagonists/pharmacology , Gene Expression Regulation/drug effects , Multidrug Resistance-Associated Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , RNA, Messenger/metabolism , Risperidone/pharmacology , ATP-Binding Cassette Transporters/genetics , Animals , Dose-Response Relationship, Drug , Glucose/pharmacology , Multidrug Resistance-Associated Proteins/genetics , Nerve Tissue Proteins , PC12 Cells/drug effects , Potassium Channels, Inwardly Rectifying , Potassium Channels, Tandem Pore Domain/genetics , Rats , Receptors, Drug , Sulfonylurea Receptors
18.
Biochim Biophys Acta ; 1670(3): 181-98, 2004 Feb 24.
Article in English | MEDLINE | ID: mdl-14980445

ABSTRACT

Tetrahydrobiopterin is an essential cofactor for nitric oxide synthase (NOS). This study was undertaken to examine the effects of intraperitoneally injected lipopolysaccharide on tetrahydrobiopterin biosynthesis in murine white and brown adipose tissues. Tetrahydrobiopterin content, catalytic activity and mRNA expression level of GTP cyclohydrolase I (GCH), rate-controlling enzyme in de novo biosynthesis of tetrahydrobiopterin, in both adipose tissues were up-regulated by 500-microg lipopolysaccharide at 6 h after the injection. On the contrary, treatment of 3T3-L1 adipocytes with lipopolysaccharide alone did not affect GCH mRNA expression level, whereas the combination of lipopolysaccharide, tumor necrosis factor (TNF)-alpha, and interferon gamma induced the increase in expression levels of GCH mRNA and CD14 mRNA. Collectively, our results showed that tetrahydrobiopterin biosynthesis can be augmented by increased GCH activity caused by a synergistic effect of lipopolysaccharide and cytokines in white and brown adipose tissues. These observations support the view that tetrahydrobiopterin biosynthesis in the adipose tissues is a target of inflammatory events triggered by peripheral LPS injection.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue/drug effects , Biopterins/analogs & derivatives , Biopterins/biosynthesis , Lipopolysaccharides/pharmacology , 3T3-L1 Cells/drug effects , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Animals , Catalysis , Cell Differentiation , Endotoxemia/chemically induced , Endotoxemia/metabolism , GTP Cyclohydrolase/biosynthesis , GTP Cyclohydrolase/genetics , GTP Cyclohydrolase/metabolism , Immunoblotting , Injections, Intraperitoneal , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/administration & dosage , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C3H , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , RNA, Messenger/analysis , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Toll-Like Receptors , Up-Regulation
19.
Brain Res ; 1039(1-2): 116-29, 2005 Mar 28.
Article in English | MEDLINE | ID: mdl-15781053

ABSTRACT

The olfactory bulb (OB) is one of the few structures in the adult mammalian CNS that contains a continuous supply of newly generated neurons in the subventricular zone. Therefore, the balance between the supply of new cells and apoptosis in the OB might determine olfactory function. Lipopolysaccharide-induced tumor necrosis factor (TNF)-alpha triggers the apoptotic cascade mediated by the TNF/TNF receptor (TNFR) pathway. The present study therefore examines the effect of the propagated innate immune reaction triggered by peripheral lipopolysaccharide on the OB of C3H/HeN mice. Within 2 h of an intraperitoneal injection of lipopolysaccharide, mRNA expression levels of the genes encoding IkappaB, TNF-alpha, and TNFR type 1 in the mouse OB were significantly enhanced. Double immunofluorescence microscopy confirmed that almost all TNF-alpha-immunopositive cells in the OB of the TNF-injected mice were located in the subependymal zone and that they overlapped cells immunostained with antibody against glial fibrillary acidic protein, but not with the antibody against F4/80, an antigenic marker of microglia. The number of TUNEL-positive cells identified exclusively in the granule cell layer was significantly increased in mice injected with lipopolysaccharide and sacrificed at 24 h thereafter. These results suggest that peripheral lipopolysaccharide causes disequilibrium between the supply and disappearance of the cells in the OB, which might lead to olfactory dysfunction.


Subject(s)
Apoptosis/immunology , Lipopolysaccharides/immunology , Olfactory Bulb/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , I-kappa B Kinase , Immunohistochemistry , Injections, Intraperitoneal , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred C3H , Olfactory Bulb/cytology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/analysis , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/immunology , Tissue Distribution , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Adv Pharmacol ; 68: 3-11, 2013.
Article in English | MEDLINE | ID: mdl-24054137

ABSTRACT

Tyrosine hydroxylase (TH), the rate-limiting enzyme in the biosynthesis of catecholamines, is a key protein involved in the pathogenesis of neurodegenerative diseases such as Parkinson's disease. Elucidation of the mechanisms regulating the synthesis, degradation, and activity of TH should be a first target in order to understand the role of this enzyme in pathogenesis. Recently, several reports suggest that the ubiquitin-proteasome pathway is a prerequisite for the degradation of TH and that the N-terminal part of TH plays a critical role in the degradation. In this report, we propose the mechanism by which the N-terminal part of TH regulates the degradation of this enzyme. Moreover, we integrate our findings with recent progress in other areas of TH regulation.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Catecholamines/metabolism , Humans , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL